1
|
Huang S, Riley AL. Drug discrimination learning: Interoceptive stimulus control of behavior and its implications for regulated and dysregulated drug intake. Pharmacol Biochem Behav 2024; 244:173848. [PMID: 39137873 DOI: 10.1016/j.pbb.2024.173848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
Drug discrimination research has generated rich evidence for the capacity of interoceptive drug stimuli to control behavior by serving as discriminative cues. Owing to its neuropharmacological specificity, drug discrimination learning has been widely used to characterize the stimulus effects and neuropharmacological underpinning of drugs. Apart from such utility, discriminative drug stimuli may help regulate drug use by disambiguating conditioned associations and post-intake outcomes. First, this review summarizes the evidence supporting interoceptive regulation of drug intake from the literature of exteroceptive discriminative control of drug-related behavior, effects of drug priming, and self-titration of drug intake. Second, an overview of interoceptive control of reward-seeking and the animal model of discriminated goal-tracking is provided to illustrate interoceptive stimulus control of the initiation and patterning of drug intake. Third, we highlight the importance of interoceptive control of aversion-avoidance in the termination of drug-use episodes and describe the animal model of discriminated taste avoidance that supports such a position. In bridging these discriminative functions of drug stimuli, we propose that interoceptive drug stimuli help regulate intake by disambiguating whether intake will be rewarding, nonrewarding, or aversive. The reflection and discussion on current theoretical formulations of interoceptive control of drug intake may further scientific advances to improve animal models to study the mechanisms by which interoceptive stimuli regulate drug intake, as well as how alterations of interoceptive processes may contribute to the transition to dysregulated drug use.
Collapse
Affiliation(s)
- Shihui Huang
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave, NW, Washington, DC 20016, USA.
| | - Anthony L Riley
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave, NW, Washington, DC 20016, USA.
| |
Collapse
|
2
|
Radnai L, Young EJ, Kikuti C, Hafenbreidel M, Stremel RF, Lin L, Toth K, Pasetto P, Jin X, Patel A, Conlon M, Briggs S, Heidsieck L, Sweeney HL, Sellers J, Krieger-Burke T, Martin WH, Sisco J, Young S, Pearson P, Rumbaugh G, Araldi GL, Duddy SK, Cameron MD, Surman M, Houdusse A, Griffin PR, Kamenecka TM, Miller CA. Development of Clinically Viable Non-Muscle Myosin II Small Molecule Inhibitors with Broad Therapeutic Potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617018. [PMID: 39416074 PMCID: PMC11482808 DOI: 10.1101/2024.10.07.617018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Non-muscle myosin II (NMII), a molecular motor that regulates critical processes such as cytokinesis and neuronal synaptic plasticity, has substantial therapeutic potential. However, translating this potential to in vivo use has been hampered by the lack of selective tools. The most prototypical non-selective inhibitor, blebbistatin inactivates both NMII and cardiac myosin II (CMII), a key regulator of heart function. Using rational drug design, we developed a series of NMII inhibitors that improve tolerability by selectively targeting NMII over CMII, including MT-228, which has excellent properties such as high brain penetration and efficacy in preclinical models of stimulant use disorder, which has no current FDA-approved therapies. The structure of MT-228 bound to myosin II provides insight into its 17-fold selectivity for NMII over CMII. MT-228's broad therapeutic window opens the door to new disease treatments and provides valuable tools for the scientific community, along with promising leads for future medication development. Highlights Research suggests numerous indications, from axon regeneration and cancer, would benefit from a small molecule inhibitor of non-muscle myosin II, a molecular motor that regulates the actin cytoskeleton. Current chemical probe options are very limited and lack sufficient safety for in vivo studies, which we show is primarily due to potent inhibition of cardiac myosin II.Rational design that focused on improving target selectivity over the pan-myosin II inhibitor, blebbistatin, led to the identification of MT-228, a small molecule inhibitor with a wide therapeutic window.High-resolution structure of MT-228 bound to myosin II reveals that selectivity results from a different positioning compared to blebbistatin and an important sequence difference between cardiac and non-muscle myosin II in the inhibitor binding pocket.A single administration of MT-228 shows long-lasting efficacy in animal models of stimulant use disorder, a current unmet and rapidly escalating need with no FDA-approved treatments.
Collapse
|
3
|
Renda B, Leri F. The anxiogenic drug yohimbine is a reinforcer in male and female rats. Neuropsychopharmacology 2024:10.1038/s41386-024-01985-1. [PMID: 39289489 DOI: 10.1038/s41386-024-01985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
The indole alkaloid yohimbine is an anxiogenic drug that activates stress-responsive systems in the brain. However, because yohimbine also elicits approach behaviors, this study employed male and female Sprague-Dawley rats to explore its potential reinforcing effects. Thus, it was first determined if intravenous (IV) infusions of yohimbine (0.25 mg/kg/infusion) could maintain lever pressing, whether intake could be modulated by dose/infusion, and if lever pressing would persist in the absence of yohimbine or yohimbine-paired cues. Next, to assess yohimbine's effect on memory consolidation, 0.3, 1.25 or 3 mg/kg yohimbine was administered post-training using an object recognition memory task. Finally, place conditioning assessed whether doses of yohimbine that elevate blood serum corticosterone levels (1.25 or 3 mg/kg) could elicit a conditioned place preference. It was found that both sexes acquired yohimbine IV self-administration, that intake was modulated by dose/infusion, and that lever pressing persisted during extinction and in the absence of the yohimbine-paired cue. As well, post-training injections of 1.25 mg/kg yohimbine enhanced consolidation of object memory, and 1.25 and 3 mg/kg elevated corticosterone levels and elicited a place preference in both sexes. Finally, in behavioral tests of psychomotor functions, acute yohimbine increased lever pressing for a visual cue and elevated locomotor activity. These findings reveal a profile of yohimbine's behavioral effects that is consistent with that of psychostimulant reinforcing drugs.
Collapse
Affiliation(s)
- Briana Renda
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Francesco Leri
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada.
- Department of Psychology, University of Toronto, Scarborough, ON, Canada.
| |
Collapse
|
4
|
Oliva HNP, Prudente TP, Nunes EJ, Cosgrove KP, Radhakrishnan R, Potenza MN, Angarita GA. Substance use and spine density: a systematic review and meta-analysis of preclinical studies. Mol Psychiatry 2024; 29:2873-2885. [PMID: 38561468 DOI: 10.1038/s41380-024-02519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
The elucidation of synaptic density changes provides valuable insights into the underlying brain mechanisms of substance use. In preclinical studies, synaptic density markers, like spine density, are altered by substances of abuse (e.g., alcohol, amphetamine, cannabis, cocaine, opioids, nicotine). These changes could be linked to phenomena including behavioral sensitization and drug self-administration in rodents. However, studies have produced heterogeneous results for spine density across substances and brain regions. Identifying patterns will inform translational studies given tools that now exist to measure in vivo synaptic density in humans. We performed a meta-analysis of preclinical studies to identify consistent findings across studies. PubMed, ScienceDirect, Scopus, and EBSCO were searched between September 2022 and September 2023, based on a protocol (PROSPERO: CRD42022354006). We screened 6083 publications and included 70 for meta-analysis. The meta-analysis revealed drug-specific patterns in spine density changes. Hippocampal spine density increased after amphetamine. Amphetamine, cocaine, and nicotine increased spine density in the nucleus accumbens. Alcohol and amphetamine increased, and cannabis reduced, spine density in the prefrontal cortex. There was no convergence of findings for morphine's effects. The effects of cocaine on the prefrontal cortex presented contrasting results compared to human studies, warranting further investigation. Publication bias was small for alcohol or morphine and substantial for the other substances. Heterogeneity was moderate-to-high across all substances. Nonetheless, these findings inform current translational efforts examining spine density in humans with substance use disorders.
Collapse
Affiliation(s)
- Henrique Nunes Pereira Oliva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Tiago Paiva Prudente
- Faculdade de Medicina, Universidade Federal de Goiás (UFG), Goiânia, Goiás, Brazil
| | - Eric J Nunes
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Yale Tobacco Center of Regulatory Science, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
- Connecticut Council on Problem Gambling, Wethersfield, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA.
| |
Collapse
|
5
|
Heal DJ, Gosden J, Smith SL. A critical assessment of the abuse, dependence and associated safety risks of naturally occurring and synthetic cannabinoids. Front Psychiatry 2024; 15:1322434. [PMID: 38915848 PMCID: PMC11194422 DOI: 10.3389/fpsyt.2024.1322434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/27/2024] [Indexed: 06/26/2024] Open
Abstract
Various countries and US States have legalized cannabis, and the use of the psychoactive1 and non-psychoactive cannabinoids is steadily increasing. In this review, we have collated evidence from published non-clinical and clinical sources to evaluate the abuse, dependence and associated safety risks of the individual cannabinoids present in cannabis. As context, we also evaluated various synthetic cannabinoids. The evidence shows that delta-9 tetrahydrocannabinol (Δ9-THC) and other psychoactive cannabinoids in cannabis have moderate reinforcing effects. Although they rapidly induce pharmacological tolerance, the withdrawal syndrome produced by the psychoactive cannabinoids in cannabis is of moderate severity and lasts from 2 to 6 days. The evidence overwhelmingly shows that non-psychoactive cannabinoids do not produce intoxicating, cognitive or rewarding properties in humans. There has been much speculation whether cannabidiol (CBD) influences the psychoactive and potentially harmful effects of Δ9-THC. Although most non-clinical and clinical investigations have shown that CBD does not attenuate the CNS effects of Δ9-THC or synthetic psychoactive cannabinoids, there is sufficient uncertainty to warrant further research. Based on the analysis, our assessment is cannabis has moderate levels of abuse and dependence risk. While the risks and harms are substantially lower than those posed by many illegal and legal substances of abuse, including tobacco and alcohol, they are far from negligible. In contrast, potent synthetic cannabinoid (CB1/CB2) receptor agonists are more reinforcing and highly intoxicating and pose a substantial risk for abuse and harm. 1 "Psychoactive" is defined as a substance that when taken or administered affects mental processes, e.g., perception, consciousness, cognition or mood and emotions.
Collapse
Affiliation(s)
- David J. Heal
- DevelRx Limited, Nottingham, United Kingdom
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | | | | |
Collapse
|
6
|
Markovic T, Higginbotham J, Ruyle B, Massaly N, Yoon HJ, Kuo CC, Kim JR, Yi J, Garcia JJ, Sze E, Abt J, Teich RH, Dearman JJ, McCall JG, Morón JA. A locus coeruleus to dorsal hippocampus pathway mediates cue-induced reinstatement of opioid self-administration in male and female rats. Neuropsychopharmacology 2024; 49:915-923. [PMID: 38374364 PMCID: PMC11039689 DOI: 10.1038/s41386-024-01828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
Opioid use disorder is a chronic relapsing disorder encompassing misuse, dependence, and addiction to opioid drugs. Long term maintenance of associations between the reinforcing effects of the drug and the cues associated with its intake are a leading cause of relapse. Indeed, exposure to the salient drug-associated cues can lead to drug cravings and drug seeking behavior. The dorsal hippocampus (dHPC) and locus coeruleus (LC) have emerged as important structures for linking the subjective rewarding effects of opioids with environmental cues. However, their role in cue-induced reinstatement of opioid use remains to be further elucidated. In this study, we showed that chemogenetic inhibition of excitatory dHPC neurons during re-exposure to drug-associated cues significantly attenuates cue-induced reinstatement of morphine-seeking behavior. In addition, the same manipulation reduced reinstatement of sucrose-seeking behavior but failed to alter memory recall in the object location task. Finally, intact activity of tyrosine hydroxylase (TH) LC-dHPCTh afferents is necessary to drive cue induced reinstatement of morphine-seeking as inhibition of this pathway blunts cue-induced drug-seeking behavior. Altogether, these studies show an important role of the dHPC and LC-dHPCTh pathway in mediating cue-induced reinstatement of opioid seeking.
Collapse
Affiliation(s)
- Tamara Markovic
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica Higginbotham
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian Ruyle
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Hye Jean Yoon
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jenny R Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jiwon Yi
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeniffer J Garcia
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric Sze
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Julian Abt
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Rachel H Teich
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Joanna J Dearman
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jose A Morón
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA.
- Pain Center, Washington University in St Louis, St. Louis, MO, USA.
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA.
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
7
|
Childs JE, Morabito S, Das S, Santelli C, Pham V, Kusche K, Vera VA, Reese F, Campbell RR, Matheos DP, Swarup V, Wood MA. Relapse to cocaine seeking is regulated by medial habenula NR4A2/NURR1 in mice. Cell Rep 2024; 43:113956. [PMID: 38489267 PMCID: PMC11100346 DOI: 10.1016/j.celrep.2024.113956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 09/11/2023] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
Drugs of abuse can persistently change the reward circuit in ways that contribute to relapse behavior, partly via mechanisms that regulate chromatin structure and function. Nuclear orphan receptor subfamily4 groupA member2 (NR4A2, also known as NURR1) is an important effector of histone deacetylase 3 (HDAC3)-dependent mechanisms in persistent memory processes and is highly expressed in the medial habenula (MHb), a region that regulates nicotine-associated behaviors. Here, expressing the Nr4a2 dominant negative (Nurr2c) in the MHb blocks reinstatement of cocaine seeking in mice. We use single-nucleus transcriptomics to characterize the molecular cascade following Nr4a2 manipulation, revealing changes in transcriptional networks related to addiction, neuroplasticity, and GABAergic and glutamatergic signaling. The network controlled by NR4A2 is characterized using a transcription factor regulatory network inference algorithm. These results identify the MHb as a pivotal regulator of relapse behavior and demonstrate the importance of NR4A2 as a key mechanism driving the MHb component of relapse.
Collapse
Affiliation(s)
- Jessica E Childs
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Samuel Morabito
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA 92697, USA; Mathematical, Computational, and Systems Biology (MCSB) Program, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Sudeshna Das
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA 92697, USA
| | - Caterina Santelli
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Victoria Pham
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Kelly Kusche
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Vanessa Alizo Vera
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Fairlie Reese
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Rianne R Campbell
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Dina P Matheos
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA 92697, USA.
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA; UC Irvine Center for Addiction Neuroscience, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
8
|
Levinstein MR, De Oliveira PA, Casajuana-Martin N, Quiroz C, Budinich RC, Rais R, Rea W, Ventriglia EN, Llopart N, Casadó-Anguera V, Moreno E, Walther D, Glatfelter GC, Weinshenker D, Zarate CA, Casadó V, Baumann MH, Pardo L, Ferré S, Michaelides M. Unique pharmacodynamic properties and low abuse liability of the µ-opioid receptor ligand (S)-methadone. Mol Psychiatry 2024; 29:624-632. [PMID: 38145984 PMCID: PMC11221360 DOI: 10.1038/s41380-023-02353-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/27/2023]
Abstract
(R,S)-methadone ((R,S)-MTD) is a µ-opioid receptor (MOR) agonist comprised of (R)-MTD and (S)-MTD enantiomers. (S)-MTD is being developed as an antidepressant and is considered an N-methyl-D-aspartate receptor (NMDAR) antagonist. We compared the pharmacology of (R)-MTD and (S)-MTD and found they bind to MORs, but not NMDARs, and induce full analgesia. Unlike (R)-MTD, (S)-MTD was a weak reinforcer that failed to affect extracellular dopamine or induce locomotor stimulation. Furthermore, (S)-MTD antagonized motor and dopamine releasing effects of (R)-MTD. (S)-MTD acted as a partial agonist at MOR, with complete loss of efficacy at the MOR-galanin Gal1 receptor (Gal1R) heteromer, a key mediator of the dopaminergic effects of opioids. In sum, we report novel and unique pharmacodynamic properties of (S)-MTD that are relevant to its potential mechanism of action and therapeutic use. One-sentence summary: (S)-MTD, like (R)-MTD, binds to and activates MORs in vitro, but (S)-MTD antagonizes the MOR-Gal1R heteromer, decreasing its abuse liability.
Collapse
Affiliation(s)
- Marjorie R Levinstein
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Paulo A De Oliveira
- Integrative Neurobiology Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Nil Casajuana-Martin
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Universitat Autònoma Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Cesar Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Reece C Budinich
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Neurology and Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - William Rea
- Integrative Neurobiology Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Emilya N Ventriglia
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Natàlia Llopart
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institut de Biomedicina de la Universitat de Barcelona, 08028, Barcelona, Spain
| | - Verònica Casadó-Anguera
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institut de Biomedicina de la Universitat de Barcelona, 08028, Barcelona, Spain
| | - Estefanía Moreno
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institut de Biomedicina de la Universitat de Barcelona, 08028, Barcelona, Spain
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Grant C Glatfelter
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health Intramural Research Program, Bethesda, MD, 20892, USA
| | - Vicent Casadó
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institut de Biomedicina de la Universitat de Barcelona, 08028, Barcelona, Spain
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Universitat Autònoma Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA.
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
9
|
Zell V, Teuns G, Needham AS, Mukherjee S, Roscoe N, Le M, Fourgeaud L, Woodruff G, Bhattacharya A, Marella M, Bonaventure P, Drevets WC, Balana B. Characterization of Selective M 5 Acetylcholine Muscarinic Receptor Modulators on Dopamine Signaling in the Striatum. J Pharmacol Exp Ther 2023; 387:226-234. [PMID: 37679045 DOI: 10.1124/jpet.123.001737] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The type-5 muscarinic acetylcholine receptor (mAChR, M5) is almost exclusively expressed in dopamine (DA) neurons of the ventral tegmental area and substantia nigra pars compacta; therefore, they are ideally located to modulate DA signaling and underlying behaviors. However, the role of M5 in shaping DA release is still poorly characterized. In this study, we first quantitatively mapped the expression of M5 in different neurons of the mouse midbrain, then used voltammetry in mouse striatum to evaluate the effect of M5-selective modulators on DA release. The M5 negative allosteric modulator ML375 significantly decreased electrically evoked DA release and blocked the effect of Oxotremorine-M (Oxo-M; nonselective mAChR agonist) on DA release in the presence of an acetylcholine nicotinic receptor blocker. Conversely, the M5 positive allosteric modulator VU 0365114 significantly increased electrically evoked DA release and the Oxo-M effect on DA release. We then assessed M5's impact on mesolimbic circuit function in vivo. Although psychostimulant-induced locomotor activity models in knockout mice have previously been used to characterize the role of M5 in DA transmission, the results of these studies conflict, leading us to select a different in vivo model, namely a cocaine self-administration paradigm. In contrast to a previous study that also used this model, in the current study, administration of ML375 did not decrease cocaine self-administration in rats (using fixed and progressive ratio). These conflicting results illustrate the complexity of M5 modulation and the need to further characterize its involvement in the regulation of dopamine signaling, central to multiple neuropsychiatric diseases. SIGNIFICANCE STATEMENT: This work describes the type-5 muscarinic receptor (M5) pattern of expression within the midbrain as well as its physiological modulation by selective compounds at the axon terminal level in the striatum, where M5 directly shapes dopamine transmission. It offers the first direct readout of mesolimbic dopamine release modulation by M5, highlighting its role in regulating neurocircuits implicated in the pathophysiology of neuropsychiatric disorders such as substance use disorders, major depressive disorder, and schizophrenia.
Collapse
Affiliation(s)
- Vivien Zell
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Greetje Teuns
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Alexandra Stormy Needham
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Sruti Mukherjee
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Nathaniel Roscoe
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Michelle Le
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Lawrence Fourgeaud
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Grace Woodruff
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Anindya Bhattacharya
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Mathieu Marella
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Pascal Bonaventure
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Wayne C Drevets
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Bartosz Balana
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| |
Collapse
|
10
|
Bagley JR, Tan Y, Zhu W, Cheng Z, Takeda S, Fang Z, Arslan A, Wang M, Guan Y, Jiang L, Jian R, Gu F, Parada I, Prince D, Jentsch JD, Peltz G. Neuron Navigator 1 (Nav1) regulates the response to cocaine in mice. Commun Biol 2023; 6:1053. [PMID: 37853211 PMCID: PMC10584906 DOI: 10.1038/s42003-023-05430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
Genetic variation accounts for much of the risk for developing a substance use disorder, but the underlying genetic factors and their genetic effector mechanisms are mostly unknown. Inbred mouse strains exhibit substantial and heritable differences in the extent of voluntary cocaine self-administration. Computational genetic analysis of cocaine self-administration data obtained from twenty-one inbred strains identified Nav1, a member of the neuron navigator family that regulates dendrite formation and axonal guidance, as a candidate gene. To test this genetic hypothesis, we generated and characterized Nav1 knockout mice. Consistent with the genetic prediction, Nav1 knockout mice exhibited increased voluntary cocaine intake and had increased motivation for cocaine consumption. Immunohistochemistry, electrophysiology, and transcriptomic studies were performed as a starting point for investigating the mechanism for the Nav1 knockout effect. Nav1 knockout mice had a reduced inhibitory synapse density in their cortex, increased excitatory synaptic transmission in their cortex and hippocampus, and increased excitatory neurons in a deep cortical layer. Collectively, our results indicate that Nav1 regulates the response to cocaine, and we identified Nav1 knockout induced changes in the excitatory and inhibitory synaptic balance in the cortex and hippocampus that could contribute to this effect.
Collapse
Affiliation(s)
- Jared R Bagley
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Yalun Tan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Wan Zhu
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Zhuanfen Cheng
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Saori Takeda
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Zhouqing Fang
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Ahmed Arslan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Meiyue Wang
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Yuan Guan
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University Medical School, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University Medical School, Stanford, CA, USA
| | - Feng Gu
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
- Department of Biological Sciences, University of North Texas, Denton, USA
| | - Isabel Parada
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
| | - David Prince
- Department of Neurology, Stanford University Medical School, Stanford, CA, USA
| | - J David Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Gary Peltz
- Department of Anesthesiology, Pain and Perioperative Medicine Stanford University Medical School, Stanford, CA, USA.
| |
Collapse
|
11
|
Salinsky LM, Merritt CR, Zamora JC, Giacomini JL, Anastasio NC, Cunningham KA. μ-opioid receptor agonists and psychedelics: pharmacological opportunities and challenges. Front Pharmacol 2023; 14:1239159. [PMID: 37886127 PMCID: PMC10598667 DOI: 10.3389/fphar.2023.1239159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
Opioid misuse and opioid-involved overdose deaths are a massive public health problem involving the intertwined misuse of prescription opioids for pain management with the emergence of extremely potent fentanyl derivatives, sold as standalone products or adulterants in counterfeit prescription opioids or heroin. The incidence of repeated opioid overdose events indicates a problematic use pattern consistent with the development of the medical condition of opioid use disorder (OUD). Prescription and illicit opioids reduce pain perception by activating µ-opioid receptors (MOR) localized to the central nervous system (CNS). Dysregulation of meso-corticolimbic circuitry that subserves reward and adaptive behaviors is fundamentally involved in the progressive behavioral changes that promote and are consequent to OUD. Although opioid-induced analgesia and the rewarding effects of abused opioids are primarily mediated through MOR activation, serotonin (5-HT) is an important contributor to the pharmacology of opioid abused drugs (including heroin and prescription opioids) and OUD. There is a recent resurgence of interest into psychedelic compounds that act primarily through the 5-HT2A receptor (5-HT 2A R) as a new frontier in combatting such diseases (e.g., depression, anxiety, and substance use disorders). Emerging data suggest that the MOR and 5-HT2AR crosstalk at the cellular level and within key nodes of OUD circuitry, highlighting a major opportunity for novel pharmacological intervention for OUD. There is an important gap in the preclinical profiling of psychedelic 5-HT2AR agonists in OUD models. Further, as these molecules carry risks, additional analyses of the profiles of non-hallucinogenic 5-HT2AR agonists and/or 5-HT2AR positive allosteric modulators may provide a new pathway for 5-HT2AR therapeutics. In this review, we discuss the opportunities and challenges associated with utilizing 5-HT2AR agonists as therapeutics for OUD.
Collapse
Affiliation(s)
| | | | | | | | - Noelle C. Anastasio
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Kathryn A. Cunningham
- Center for Addiction Sciences and Therapeutics and Department of Pharmacology and Toxicology, John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
12
|
Luján MÁ, Oliver BL, Young-Morrison R, Engi SA, Zhang LY, Wenzel JM, Li Y, Zlebnik NE, Cheer JF. A multivariate regressor of patterned dopamine release predicts relapse to cocaine. Cell Rep 2023; 42:112553. [PMID: 37224011 DOI: 10.1016/j.celrep.2023.112553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/28/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
Understanding mesolimbic dopamine adaptations underlying vulnerability to drug relapse is essential to inform prognostic tools for effective treatment strategies. However, technical limitations have hindered the direct measurement of sub-second dopamine release in vivo for prolonged periods of time, making it difficult to gauge the weight that these dopamine abnormalities have in determining future relapse incidence. Here, we use the fluorescent sensor GrabDA to record, with millisecond resolution, every single cocaine-evoked dopamine transient in the nucleus accumbens (NAc) of freely moving mice during self-administration. We reveal low-dimensional features of patterned dopamine release that are strong predictors of cue-induced reinstatement of cocaine seeking. Additionally, we report sex-specific differences in cocaine-related dopamine responses related to a greater resistance to extinction in males compared with females. These findings provide important insights into the sufficiency of NAc dopamine signaling dynamics-in interaction with sex-for recapitulating persistent cocaine seeking and future relapse vulnerability.
Collapse
Affiliation(s)
- Miguel Á Luján
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brandon L Oliver
- Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA, USA
| | - Reana Young-Morrison
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sheila A Engi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lan-Yuan Zhang
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jennifer M Wenzel
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Natalie E Zlebnik
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA, USA.
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
Sapko MT, Hanania T, Chang Q, Javitt JC. D-cycloserine is not susceptible to self-administration using an intravenous self-administration model in male ketamine-habituated Sprague-Dawley rats. Pharmacol Biochem Behav 2023:173586. [PMID: 37330114 DOI: 10.1016/j.pbb.2023.173586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
OBJECTIVE N-methyl-d-aspartate receptor (NMDAR) antagonist antidepressants have known potential for abuse liability. The aim of this study was to evaluate the abuse liability of D-cycloserine (DCS), using a self-administration paradigm in which DCS was tested for its efficacy in substituting for ketamine in ketamine-dependent rats. METHODS A standard intravenous self-administration study was conducted in male adult Sprague-Dawley rats to study compounds' abuse liability. Potential for self-administration was assessed in ketamine-habituated subjects. Subjects were trained to press a lever to obtain food, prior to connection of the lever to the intravenous drug administration apparatus. DCS was provided for self-infusion by test subjects at doses of 1.5, 5.0, and 15 mg/kg per lever press. RESULTS S-ketamine was seen to substitute for ketamine and to result in self-administration at the same frequency. DCS was not seen to result in self-administration at any of the test doses. The self-infusion behavior of DCS was similar to control (saline). CONCLUSION D-cycloserine, a partial agonist of the NMDAR glycine site, which has been shown to have antidepressant and anti-suicidal properties in clinical studies, has no apparent potential for abuse liability in a standard rodent self-administration model.
Collapse
Affiliation(s)
- Michael T Sapko
- NRx Pharmaceuticals, Inc., Wilmington, DE, United States of America
| | - Taleen Hanania
- PsychoGenics Inc., 215 College Road, Paramus, NJ, United States of America
| | - Qing Chang
- PsychoGenics Inc., 215 College Road, Paramus, NJ, United States of America
| | - Jonathan C Javitt
- NRx Pharmaceuticals, Inc., Wilmington, DE, United States of America; Johns Hopkins University School of Medicine (Baltimore, MD), United States of America.
| |
Collapse
|
14
|
Ventriglia E, Rizzo A, Gomez JL, Friedman J, Lam S, Solís O, Rais R, Bonaventura J, Michaelides M. Essential role of P-glycoprotein in the mechanism of action of oliceridine. Neuropsychopharmacology 2023; 48:831-842. [PMID: 36434081 PMCID: PMC10066384 DOI: 10.1038/s41386-022-01507-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/26/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022]
Abstract
Mu opioid receptor (MOR) agonists comprise the most effective analgesics, but their therapeutic utility is limited by adverse effects. One approach for limiting such effects has been to develop "biased" MOR agonists that show preference for activating G protein over β-Arrestin signaling. However, the notion of biased agonism has been challenged by recent studies. Oliceridine (Olinvyk®, TRV-130, OLC) is a selective MOR agonist approved by the FDA in 2020 for pain management in controlled clinical settings. Oliceridine purportedly demonstrates diminished adverse effects compared to morphine or other MOR agonists, a profile attributed to its biased agonism. However, recent studies suggest that oliceridine does not display biased agonism but instead weak intrinsic efficacy for G protein and β-Arrestin activation. Nevertheless, these insights have been derived from in vitro studies. To better understand oliceridine's in vivo efficacy profile, we performed a comprehensive assessment of its in vitro and in vivo pharmacology using both cultured cells and rodents. In vitro, oliceridine displayed high MOR affinity and weak intrinsic efficacy. In vivo, oliceridine showed impaired brain penetrance and rapid clearance, effects we attributed to its interaction with the P-glycoprotein (P-gp) efflux transporter. Moreover, we found that P-gp was essential for oliceridine's in vivo efficacy and adverse effect profiles. Taken together with prior studies, our results suggest that oliceridine's in vivo efficacy and adverse effect profiles are not attributed solely to its weak intrinsic efficacy or biased agonism but, to a large extent, its interaction with P-gp as well.
Collapse
Affiliation(s)
- Emilya Ventriglia
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Arianna Rizzo
- Departament de Patologia i Terapèutica Experimental, Institute de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Jacob Friedman
- Medications Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Sherry Lam
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Oscar Solís
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
- Departament de Patologia i Terapèutica Experimental, Institute de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain.
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain.
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
15
|
Moline M, Asakura S, Beuckman C, Landry I, Setnik B, Ashworth J, Henningfield JE. The abuse potential of lemborexant, a dual orexin receptor antagonist, according to the 8 factors of the Controlled Substances Act. Psychopharmacology (Berl) 2023; 240:699-711. [PMID: 36749354 PMCID: PMC10006052 DOI: 10.1007/s00213-023-06320-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/17/2023] [Indexed: 02/08/2023]
Abstract
RATIONALE Lemborexant (LEM) is a dual orexin receptor antagonist (DORA) approved in multiple countries including the USA, Japan, Canada, Australia, and several Asian countries for the treatment of insomnia in adults. As a compound with central nervous system activity, it is important to understand the abuse potential of LEM with respect to public health. OBJECTIVES This review discusses data for LEM relevant to each of the 8 factors of the United States Controlled Substances Act. RESULTS LEM did not demonstrate abuse potential in nonclinical testing and was associated with a low incidence of abuse-related adverse events in clinical study participants with insomnia disorder. Similar to other DORAs that have been evaluated (eg., almorexant, suvorexant (SUV), and daridorexant), LEM and the positive controls (zolpidem and SUV) also showed drug liking in a phase 1 abuse potential study that enrolled subjects who used sedatives recreationally. However, internet surveillance of SUV and the FDA Adverse Events Reporting System suggests that drugs in the DORA class display very low abuse-related risks in the community. Additionally, as described in FDA-approved labeling, it does not carry physical dependence and withdrawal risks. CONCLUSIONS LEM, similar to most other prescription insomnia medications, was placed into Schedule IV. However, LEM and other drugs in the DORA class may have a lower potential for abuse as suggested by real-world postmarketing data from federal surveys and internet surveillance, and thus may have lower risks to public health than Schedule IV benzodiazepines and nonbenzodiazepine hypnotics that potentiate GABA signaling.
Collapse
Affiliation(s)
- Margaret Moline
- Eisai Inc., 200 Metro Boulevard, Nutley, Jersey, NJ, 07110, USA.
| | | | | | | | - Beatrice Setnik
- Altasciences, Laval, Quebec, Canada and the Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | | | - Jack E Henningfield
- Pinney Associates, Inc., Bethesda, MD, USA.,The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Baird TR, Karin KN, Marsh SA, Carroll FI, Medina-Contreras JML, Negus SS, Eltit JM. Rate of onset of dopamine transporter inhibitors assessed with intracranial self-stimulation and in vivo dopamine photometry in rats. Psychopharmacology (Berl) 2023; 240:969-981. [PMID: 36802016 PMCID: PMC10466267 DOI: 10.1007/s00213-023-06340-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/09/2023] [Indexed: 02/23/2023]
Abstract
Drug self-administration and intracranial self-stimulation (ICSS) are two preclinical behavioral procedures used to predict abuse potential of drugs, and abuse-related drug effects in both procedures are thought to depend on increased mesolimbic dopamine (DA) signaling. Drug self-administration and ICSS yield concordant metrics of abuse potential across a diverse range of drug mechanisms of action. The "rate of onset," defined as the velocity with which a drug produces its effect once administered, has also been implicated as a determinant of abuse-related drug effects in self-administration procedures, but this variable has not been systematically examined in ICSS. Accordingly, this study compared ICSS effects produced in rats by three DA transporter inhibitors that have different rates of onset (fastest to slowest: cocaine, WIN-35428, RTI-31) and that produced progressively weaker metrics of abuse potential in a drug self-administration procedure in rhesus monkeys. Additionally, in vivo photometry using the fluorescent DA sensor dLight1.1 targeted to the nucleus accumbens (NAc) was used to assess the time course of extracellular DA levels as a neurochemical correlate of behavioral effects. All three compounds produced ICSS facilitation and increased DA levels assessed by dLight. In both procedures, the rank order of onset rate was cocaine > WIN-35428 > RTI-31; however, in contrast to monkey drug self-administration results, maximum effects did not differ across compounds. These results provide additional evidence that drug-induced increases in DA drive ICSS facilitation in rats and illustrate the utility of both ICSS and photometry to evaluate the time course and magnitude of abuse-related drug effects in rats.
Collapse
Affiliation(s)
- Tyson R Baird
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Integrative Life Sciences Doctoral Program, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Kimberly N Karin
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Samuel A Marsh
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - F Ivy Carroll
- Research Triangle Institute, Research Triangle Park, Durham, NC, 27709, USA
| | - J M L Medina-Contreras
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, 1101 E. Marshall Street, 3-038H, Richmond, VA, 23298, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, 1101 E. Marshall Street, 3-038H, Richmond, VA, 23298, USA.
| |
Collapse
|
17
|
Jastrzębska J, Frankowska M, Smaga I, Hubalewska-Mazgaj M, Suder A, Pieniążek R, Przegaliński E, Filip M. Evaluation of the 5-HT 2C receptor drugs RO 60-0175, WAY 161503 and mirtazepine in a preclinical model of comorbidity of depression and cocaine addiction. Pharmacol Rep 2023; 75:99-118. [PMID: 36374478 PMCID: PMC9889480 DOI: 10.1007/s43440-022-00428-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Epidemiological data indicate a high rate of comorbidity of depression and cocaine use disorder (CUD). The role of serotonin 2C (5-HT2C) receptors in the mechanisms responsible for the coexistence of depression and CUD was not investigated. METHODS We combined bilateral olfactory bulbectomy (OBX), an animal model of depression, with intravenous cocaine self-administration and extinction/reinstatement in male rats to investigate two 5-HT2C receptor agonists (Ro 60-0175 (RO) and WAY 161503 (WAY)) and the 5-HT2C-receptor preferring antagonist mirtazapine (MIR; an antidepressant), with the goal of determining whether these drugs alter cocaine-induced reinforcement and seeking behaviors. Additionally, neurochemical analyses were performed following cocaine self-administration and its abstinence period in the brain structures in OBX rats and SHAM-operated controls. RESULTS Acute administration of RO reduced, while WAY non-significantly attenuated cocaine reinforcement in both rat phenotypes. Moreover, RO or WAY protected against cocaine-seeking behavior after acute or after repeated drug administration during extinction training in OBX and SHAM rats. By contrast, acutely administered MIR did not alter cocaine reinforcement in both rat phenotypes, while it's acute (but not repeated) pretreatment reduced cocaine-seeking in OBX and SHAM rats. In neurochemical analyses, cocaine reinforcement increased 5-HT2C receptor levels in the ventral hippocampus; a preexisting depression-like phenotype enhanced this effect. The 10-daily cocaine abstinence reduced 5-HT2C receptor expression in the dorsolateral striatum, while the coexistence of depression and CUD enhanced local receptor expression. CONCLUSION The results support a key role of 5-HT2C receptors for treating CUD and comorbid depression and CUD. They may be backs the further research of pharmacological strategies with drug targeting receptors.
Collapse
Affiliation(s)
- Joanna Jastrzębska
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Małgorzata Frankowska
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Irena Smaga
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Magdalena Hubalewska-Mazgaj
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Agata Suder
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Renata Pieniążek
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Edmund Przegaliński
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Małgorzata Filip
- Maj Institute of Pharmacology, Department of Drug Addiction Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| |
Collapse
|
18
|
Pagare P, Obeng S, Huang B, Marcus MM, Nicholson KL, Townsend AE, Banks ML, Zhang Y. Preclinical Characterization and Development on NAQ as a Mu Opioid Receptor Partial Agonist for Opioid Use Disorder Treatment. ACS Pharmacol Transl Sci 2022; 5:1197-1209. [PMID: 36407950 PMCID: PMC9667545 DOI: 10.1021/acsptsci.2c00178] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Indexed: 11/06/2022]
Abstract
Mu opioid receptor (MOR) selective antagonists and partial agonists have clinical utility for the treatment of opioid use disorders (OUDs). However, the development of many has suffered due to their poor pharmacokinetic properties and/or rapid metabolism. Our recent efforts to identify MOR modulators have provided 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3-carboxamido)morphinan (NAQ), a low-efficacy partial agonist, that showed sub-nanomolar binding affinity to the MOR (K i 0.6 nM) with selectivity over the delta opioid receptor (δ/μ 241) and the kappa opioid receptor (κ/μ 48). Its potent inhibition of the analgesic effect of morphine (AD50 0.46 mg/kg) and precipitation of significantly less withdrawal symptoms even at 100-fold greater dose than naloxone represents a promising molecule for further development as a novel OUD therapeutic agent. Therefore, further in vitro and in vivo characterization of its pharmacokinetics and pharmacodynamics properties was conducted to fully understand its pharmaceutical profile. NAQ showed favorable in vitro ADMET properties and no off-target binding to several classes of GPCRs, enzymes, and ion channels. Following intravenous administration, 1 mg/kg dose of NAQ showed a similar in vivo pharmacokinetic profile to naloxone; however, orally administered 10 mg/kg NAQ demonstrated significantly improved oral bioavailability over both naloxone and naltrexone. Abuse liability assessment of NAQ in rats demonstrated that NAQ functioned as a less potent reinforcer than heroin. Chronic 5 day NAQ pretreatment decreased heroin self-administration in a heroin-vs-food choice procedure similar to the clinically used MOR partial agonist buprenorphine. Taken together, these studies provide evidence supporting NAQ as a promising lead to develop novel OUD therapeutics.
Collapse
Affiliation(s)
- Piyusha
P. Pagare
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| | - Samuel Obeng
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| | - Boshi Huang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| | - Madison M. Marcus
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Katherine L. Nicholson
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Andrew E. Townsend
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Matthew L. Banks
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University School of Medicine, Richmond, Virginia23298-0613, United States
| | - Yan Zhang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia23298-0540, United States
| |
Collapse
|
19
|
Effects of the Phenethylamine 2-Cl-4,5-MDMA and the Synthetic Cathinone 3,4-MDPHP in Adolescent Rats: Focus on Sex Differences. Biomedicines 2022; 10:biomedicines10102336. [PMID: 36289598 PMCID: PMC9598216 DOI: 10.3390/biomedicines10102336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/03/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
The illicit drug market of novel psychoactive substances (NPSs) is expanding, becoming an alarming threat due to increasing intoxication cases and insufficient (if any) knowledge of their effects. Phenethylamine 2-chloro-4,5-methylenedioxymethamphetamine (2-Cl-4,5-MDMA) and synthetic cathinone 3,4-methylenedioxy-α-pyrrolidinohexanophenone (3,4-MDPHP) are new, emerging NPSs suggested to be particularly dangerous. This study verified whether these two new drugs (i) possess abuse liability, (ii) alter plasma corticosterone levels, and (iii) interfere with dopaminergic transmission; male and female adolescent rats were included to evaluate potential sex differences in the drug-induced effects. Findings show that the two NPSs are not able to sustain reliable self-administration behavior in rats, with cumulatively earned injections of drugs being not significantly different from cumulatively earned injections of saline in control groups. Yet, at the end of the self-administration training, females (but not males) exhibited higher plasma corticosterone levels after chronic exposure to low levels of 3,4-MDPHP (but not of 2-Cl-4,5-MDMA). Finally, electrophysiological patch-clamp recordings in the rostral ventral tegmental area (rVTA) showed that both drugs are able to increase the firing rate of rVTA dopaminergic neurons in males but not in females, confirming the sex dimorphic effects of these two NPSs. Altogether, this study demonstrates that 3,4-MDPHP and 2-Cl-4,5-MDMA are unlikely to induce dependence in occasional users but can induce other effects at both central and peripheral levels that may significantly differ between males and females.
Collapse
|
20
|
Liu X, Vickstrom CR, Yu H, Liu S, Snarrenberg ST, Friedman V, Mu L, Chen B, Kelly TJ, Baker DA, Liu QS. Epac2 in midbrain dopamine neurons contributes to cocaine reinforcement via enhancement of dopamine release. eLife 2022; 11:e80747. [PMID: 35993549 PMCID: PMC9436413 DOI: 10.7554/elife.80747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/21/2022] [Indexed: 11/21/2022] Open
Abstract
Repeated exposure to drugs of abuse results in an upregulation of cAMP signaling in the mesolimbic dopamine system, a molecular adaptation thought to be critically involved in the development of drug dependence. Exchange protein directly activated by cAMP (Epac2) is a major cAMP effector abundantly expressed in the brain. However, it remains unknown whether Epac2 contributes to cocaine reinforcement. Here, we report that Epac2 in the mesolimbic dopamine system promotes cocaine reinforcement via enhancement of dopamine release. Conditional knockout of Epac2 from midbrain dopamine neurons (Epac2-cKO) and the selective Epac2 inhibitor ESI-05 decreased cocaine self-administration in mice under both fixed-ratio and progressive-ratio reinforcement schedules and across a broad range of cocaine doses. In addition, Epac2-cKO led to reduced evoked dopamine release, whereas Epac2 agonism robustly enhanced dopamine release in the nucleus accumbens in vitro. This mechanism is central to the behavioral effects of Epac2 disruption, as chemogenetic stimulation of ventral tegmental area (VTA) dopamine neurons via deschloroclozapine (DCZ)-induced activation of Gs-DREADD increased dopamine release and reversed the impairment of cocaine self-administration in Epac2-cKO mice. Conversely, chemogenetic inhibition of VTA dopamine neurons with Gi-DREADD reduced dopamine release and cocaine self-administration in wild-type mice. Epac2-mediated enhancement of dopamine release may therefore represent a novel and powerful mechanism that contributes to cocaine reinforcement.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Hao Yu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Shuai Liu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Shana Terai Snarrenberg
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Bixuan Chen
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| | - David A Baker
- Department of Biomedical Sciences, Marquette UniversityMilwaukeeUnited States
| | - Qing-song Liu
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukeeUnited States
| |
Collapse
|
21
|
Interactions between opioids and stimulants: Behavioral pharmacology of abuse-related effects. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:1-33. [PMID: 35341563 DOI: 10.1016/bs.apha.2021.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opioid abuse continues to be a significant public health challenge, with rates of opioid-related overdose deaths increasing continuously over the last two decades. There also has been a sharp increase in overdose deaths involving stimulant drugs, primarily cocaine and methamphetamine. Recent estimates indicate a high prevalence of co-use of opioids and stimulants, which is a particularly complex problem. Behavioral pharmacology research over the last few decades has characterized interactions between opioids and stimulants as well as evaluated potential treatments. This chapter describes interactions between opioids and stimulants, with a focus on pre-clinical studies of abuse-related behavioral effects using self-administration, reinstatement, drug discrimination, place conditioning, and intracranial self-stimulation paradigms in laboratory animals. In general, the literature provides substantial evidence of mutual enhancement between opioids and stimulants for abuse-related effects, although such results are not ubiquitous. Enhanced abuse-related effects could manifest in many ways including engaging in drug seeking and taking behaviors with greater persistence, effort, and motivation and/or increased likelihood of relapse. Moreover, studies on opioid/stimulant combinations set the stage for evaluating potential treatments for polysubstance use. Behavioral pharmacology research has proven invaluable for elucidating these relationships using rigorous experimental designs and quantitative analyses of pharmacological and behavioral data.
Collapse
|
22
|
Bagley JR, Khan AH, Smith DJ, Jentsch JD. Extreme phenotypic diversity in operant response to intravenous cocaine or saline infusion in the hybrid mouse diversity panel. Addict Biol 2022; 27:e13162. [PMID: 35470554 PMCID: PMC9870574 DOI: 10.1111/adb.13162] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 01/26/2023]
Abstract
Cocaine self-administration is a complexly determined trait, with a substantial proportion of individual differences being determined by genetic variation. However, the relevant genetic variants that drive heritable differences in cocaine use remain undiscovered. Cocaine intravenous self-administration (IVSA) procedures in laboratory animals provide opportunities to prospectively investigate neurogenetic influences on the acquisition of voluntary cocaine use. Here, we provide information on cocaine (or saline-as a control) IVSA in 84 members of the hybrid mouse diversity panel (HMDP), an array of genetically distinct classical or recombinant inbred strains. We found cocaine IVSA to be substantially heritable in this population, with strain-level intake ranging for near 0 to >25 mg/kg/session. Though saline IVSA was also found to be heritable, a modest genetic correlation between cocaine and saline IVSA indicates that operant responding for the cocaine reinforcer was influenced, at least in part, by unique genetic variants. Genome-wide association studies (GWAS) of infusions earned in cocaine and saline groups revealed significant quantitative trait loci (QTL) on Chromosomes 3 and 14 for cocaine, but not saline, IVSA. Positional candidates were further prioritized through use of bulk RNA sequencing data that revealed genes with cis-eQTL and genetic correlation to number of infusions. Additionally, these data identify reference strains with extreme cocaine IVSA phenotypes, revealing them as polygenic models of risk and resilience to cocaine reinforcement. This work is part of an ongoing effort to characterize genetic variation that moderates cocaine IVSA that may, in turn, provide a more comprehensive understanding of cocaine risk genetics and neurobiology.
Collapse
Affiliation(s)
- Jared R. Bagley
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Arshad H. Khan
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| | - Desmond J. Smith
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| | - James D. Jentsch
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
23
|
Riley AL, Manke HN, Huang S. Impact of the Aversive Effects of Drugs on Their Use and Abuse. Behav Neurol 2022; 2022:8634176. [PMID: 35496768 PMCID: PMC9045991 DOI: 10.1155/2022/8634176] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/16/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
Drug use and abuse are complex issues in that the basis of each may involve different determinants and consequences, and the transition from one to the other may be equally multifaceted. A recent model of the addiction cycle (as proposed by Koob and his colleagues) illustrates how drug-taking patterns transition from impulsive (acute use) to compulsive (chronic use) as a function of various neuroadaptations leading to the downregulation of DA systems, upregulation of stress systems, and the dysregulation of the prefrontal/orbitofrontal cortex. Although the nature of reinforcement in the initiation and mediation of these effects may differ (positive vs. negative), the role of reinforcement in drug intake (acute and chronic) is well characterized. However, drugs of abuse have other stimulus properties that may be important in their use and abuse. One such property is their aversive effects that limit drug intake instead of initiating and maintaining it. Evidence of such effects comes from both clinical and preclinical populations. In support of this position, the present review describes the aversive effects of drugs (assessed primarily in conditioned taste aversion learning), the fact that they occur concurrently with reward as assessed in combined taste aversion/place preference designs, the role of aversive effects in drug-taking (in balance with their rewarding effects), the dissociation of these affective properties in that they can be affected in different ways by the same manipulations, and the impact of various parametric, experiential, and subject factors on the aversive effects of drugs and the consequent impact of these factors on their use and abuse potential.
Collapse
Affiliation(s)
- Anthony L. Riley
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave NW, Washington, D.C. 20016, USA
| | - Hayley N. Manke
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave NW, Washington, D.C. 20016, USA
| | - Shihui Huang
- Psychopharmacology Laboratory, Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Ave NW, Washington, D.C. 20016, USA
| |
Collapse
|
24
|
Redish AD, Kepecs A, Anderson LM, Calvin OL, Grissom NM, Haynos AF, Heilbronner SR, Herman AB, Jacob S, Ma S, Vilares I, Vinogradov S, Walters CJ, Widge AS, Zick JL, Zilverstand A. Computational validity: using computation to translate behaviours across species. Philos Trans R Soc Lond B Biol Sci 2022; 377:20200525. [PMID: 34957854 PMCID: PMC8710889 DOI: 10.1098/rstb.2020.0525] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/28/2021] [Indexed: 11/12/2022] Open
Abstract
We propose a new conceptual framework (computational validity) for translation across species and populations based on the computational similarity between the information processing underlying parallel tasks. Translating between species depends not on the superficial similarity of the tasks presented, but rather on the computational similarity of the strategies and mechanisms that underlie those behaviours. Computational validity goes beyond construct validity by directly addressing questions of information processing. Computational validity interacts with circuit validity as computation depends on circuits, but similar computations could be accomplished by different circuits. Because different individuals may use different computations to accomplish a given task, computational validity suggests that behaviour should be understood through the subject's point of view; thus, behaviour should be characterized on an individual level rather than a task level. Tasks can constrain the computational algorithms available to a subject and the observed subtleties of that behaviour can provide information about the computations used by each individual. Computational validity has especially high relevance for the study of psychiatric disorders, given the new views of psychiatry as identifying and mediating information processing dysfunctions that may show high inter-individual variability, as well as for animal models investigating aspects of human psychiatric disorders. This article is part of the theme issue 'Systems neuroscience through the lens of evolutionary theory'.
Collapse
Affiliation(s)
- A. David Redish
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Adam Kepecs
- Department of Neuroscience, Washington University in St. Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University in St. Louis, St Louis, MO 63110, USA
| | - Lisa M. Anderson
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Olivia L. Calvin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicola M. Grissom
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ann F. Haynos
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Alexander B. Herman
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Suma Jacob
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sisi Ma
- Department of Medicine - Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Iris Vilares
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sophia Vinogradov
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cody J. Walters
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alik S. Widge
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jennifer L. Zick
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anna Zilverstand
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
25
|
Kim JM, Han B, Min HK, Yun J, Kim YH, Choi SO, Kang MS. Rewarding and reinforcing effects of two dissociative-based new psychoactive substances, deschloroketamine and diphenidine, in mice. Pharmacol Biochem Behav 2022; 213:173333. [DOI: 10.1016/j.pbb.2022.173333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 10/19/2022]
|
26
|
Hiranita T, Obeng S, Sharma A, Wilkerson JL, McCurdy CR, McMahon LR. In vitro and in vivo pharmacology of kratom. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:35-76. [PMID: 35341571 DOI: 10.1016/bs.apha.2021.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Kratom products have been historically and anecdotally used in south Asian countries for centuries to manage pain and opioid withdrawal. The use of kratom products has dramatically increased in the United States. More than 45 kratom alkaloids have been isolated, yet the overall pharmacology of the individual alkaloids is still not well characterized. The purpose of this chapter is to summarize in vitro and in vivo opioid activities of the primary kratom alkaloid mitragynine and its more potent metabolite 7-hydroxymitragynine. Following are experimental procedures described to characterize opioid receptor activity; receptor binding and functional assays, antinociceptive assays, operant conditioning assays, and respiratory plethysmography. The capacity of kratom alkaloids to confer tolerance and physical dependence as well as their pharmacokinetic properties are also summarized. The data reviewed here suggest that kratom products and mitragynine possess low efficacy agonist activity at the mu-opioid receptor in vivo. In addition, kratom products and mitragynine have been demonstrated to antagonize the effects of high efficacy mu-opioid agonists. The data further suggest that 7-hydroxymitragynine formed in vivo by metabolism of mitragynine may be minimally involved in the overall behavioral profile of mitragynine and kratom, whereas 7-hydroxymitragynine itself, at sufficiently high doses administered exogenously, shares many of the same abuse- and dependence-related behavioral effects associated with traditional opioid agonists. The apparent low efficacy of kratom products and mitragynine at mu-opioid receptors supports the development of these ligands as effective and potentially safe medications for opioid use disorder.
Collapse
Affiliation(s)
- Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Samuel Obeng
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Abhisheak Sharma
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
27
|
Negus SS, Banks ML. Confronting the challenge of failed translation in medications development for substance use disorders. Pharmacol Biochem Behav 2021; 210:173264. [PMID: 34461148 PMCID: PMC8418188 DOI: 10.1016/j.pbb.2021.173264] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/20/2021] [Indexed: 11/25/2022]
Affiliation(s)
- S S Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America.
| | - M L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States of America
| |
Collapse
|
28
|
McLaurin KA, Harris M, Madormo V, Harrod SB, Mactutus CF, Booze RM. HIV-Associated Apathy/Depression and Neurocognitive Impairments Reflect Persistent Dopamine Deficits. Cells 2021; 10:2158. [PMID: 34440928 PMCID: PMC8392364 DOI: 10.3390/cells10082158] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals living with human immunodeficiency virus type 1 (HIV-1) are often plagued by debilitating neurocognitive impairments and affective alterations;the pathophysiology underlying these deficits likely includes dopaminergic system dysfunction. The present review utilized four interrelated aims to critically examine the evidence for dopaminergic alterations following HIV-1 viral protein exposure. First, basal dopamine (DA) values are dependent upon both brain region andexperimental approach (i.e., high-performance liquid chromatography, microdialysis or fast-scan cyclic voltammetry). Second, neurochemical measurements overwhelmingly support decreased DA concentrations following chronic HIV-1 viral protein exposure. Neurocognitive impairments, including alterations in pre-attentive processes and attention, as well as apathetic behaviors, provide an additional line of evidence for dopaminergic deficits in HIV-1. Third, to date, there is no compelling evidence that combination antiretroviral therapy (cART), the primary treatment regimen for HIV-1 seropositive individuals, has any direct pharmacological action on the dopaminergic system. Fourth, the infection of microglia by HIV-1 viral proteins may mechanistically underlie the dopamine deficit observed following chronic HIV-1 viral protein exposure. An inclusive and critical evaluation of the literature, therefore, supports the fundamental conclusion that long-term HIV-1 viral protein exposure leads to a decreased dopaminergic state, which continues to persist despite the advent of cART. Thus, effective treatment of HIV-1-associated apathy/depression and neurocognitive impairments must focus on strategies for rectifying decreases in dopamine function.
Collapse
Affiliation(s)
| | | | | | | | | | - Rosemarie M. Booze
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (K.A.M.); (M.H.); (V.M.); (S.B.H.); (C.F.M.)
| |
Collapse
|
29
|
Wiley JL, Taylor SI, Marusich JA. Δ 9-Tetrahydrocannabinol discrimination: Effects of route of administration in rats. Drug Alcohol Depend 2021; 225:108827. [PMID: 34186444 PMCID: PMC8282763 DOI: 10.1016/j.drugalcdep.2021.108827] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 01/20/2023]
Abstract
Cannabis users typically smoke or vape cannabis or ingest it in edibles, whereas cannabinoids are typically administered via injection in rodent research. The present study examined the effects of route of administration (ROA) of Δ9-tetrahydrocannabinol (THC), the primary psychoactive constituent of cannabis. Adult female and male Long Evans rats were trained to discriminate intraperitoneal (i.p.) THC from vehicle in a drug discrimination procedure. Following acquisition, dose-effect curves were determined with THC using i.p., oral (p.o.), and subcutaneous (s.c.) injection in both sexes and aerosol exposure in males only, followed by a time course with one dose for each ROA. Both sexes acquired THC discrimination in a similar number of sessions, although baseline response rates were significantly lower in females than males. THC fully substituted for the 3 mg/kg i.p. training dose across all ROA. While potencies were similar for ROA involving first-pass metabolism (i.p. and p.o.), THC potency was lower with s.c. administration. During the time course analysis, aerosol administration had the shortest latency to onset of discriminative stimulus effects and the shortest duration of effect, whereas s.c. administration had the longest duration. The results of this examination of the effects of ROA on an abuse-related effect of THC provide an empirical foundation to facilitate choice of ROA for mechanistic investigation of THC's pharmacology. Further, animal models using translationally relevant ROA may facilitate more accurate predictions of their effects in humans.
Collapse
Affiliation(s)
- Jenny L Wiley
- RTI International, 3040 Cornwallis Road, Research Triangle Park, NC, 27709, USA.
| | - Shanequa I Taylor
- RTI International, 3040 Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Julie A Marusich
- RTI International, 3040 Cornwallis Road, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
30
|
Swain Y, Gewirtz JC, Harris AC. Behavioral predictors of individual differences in opioid addiction vulnerability as measured using i.v. self-administration in rats. Drug Alcohol Depend 2021; 221:108561. [PMID: 33588371 PMCID: PMC8048102 DOI: 10.1016/j.drugalcdep.2021.108561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Like other forms of psychopathology, vulnerability to opioid addiction is subject to wide individual differences. Animal behavioral models are valuable in advancing our understanding of mechanisms underlying vulnerability to the disorder's development and amenability to treatment. METHODS This review provides an overview of preclinical work on behavioral predictors of opioid addiction vulnerability as measured using the intravenous (i.v.) self-administration (SA) model in rats. We also highlight several new approaches to studying individual differences in opioid addiction vulnerability in preclinical models that could have greater sensitivity and lead to more clinically relevant findings. RESULTS AND CONCLUSIONS Evidence for the relationship between various behavioral traits and opioid SA in the preclinical literature is limited. With the possible exceptions of sensitivity to opioid agonist/withdrawal effects and stress reactivity, predictors of individual differences in SA of other drugs of abuse (e.g. sensation-seeking, impulsivity) do not predict vulnerability to opioid SA in rats. Refinement of SA measures and the use of multivariate designs and statistics could help identify predictors of opioid SA and lead to more clinically relevant studies on opioid addiction vulnerability.
Collapse
Affiliation(s)
- Yayi Swain
- Departments of Psychology, University of Minnesota, United States; Hennepin Healthcare Research Institute, United States
| | | | - Andrew C Harris
- Departments of Psychology, University of Minnesota, United States; Hennepin Healthcare Research Institute, United States; Departments of Medicine, University of Minnesota, United States.
| |
Collapse
|
31
|
Abstract
Animal models of addictive behaviors are useful for uncovering neural mechanisms involved in the development of dependence and for identifying risk factors for drug abuse. One such risk factor is biological sex, which strongly moderates drug self-administration behavior in rodents. Female rodents are more likely to acquire drug self-administration behaviors, consume higher amounts of drug, and reinstate drug-seeking behavior more readily. Despite this female vulnerability, preclinical addiction research has largely been done in male animals. The study of sex differences in rodent models of addictive behavior is increasing, however, as more investigators are choosing to include both male and female animals in experiments. This commentary is meant to serve as an introductory guide for preclinical investigators new to the study of sex differences in addiction. We provide an overview of self-administration models, a broad view of female versus male self-administration behaviors, and suggestions for study design and implementation. Inclusion of female subjects in preclinical addiction research is timely, as problem drug and alcohol use in women is increasing. With proper attention, design, and analysis, the study of sex differences in addiction has the potential to uncover novel neural mechanisms and lead to greater translational success for addiction research. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anna K. Radke
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, Ohio
| | - Elizabeth A. Sneddon
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, Ohio
| | - Sean C. Monroe
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, Ohio
| |
Collapse
|
32
|
Manduca A, Carbone E, Schiavi S, Cacchione C, Buzzelli V, Campolongo P, Trezza V. The neurochemistry of social reward during development: What have we learned from rodent models? J Neurochem 2021; 157:1408-1435. [PMID: 33569830 DOI: 10.1111/jnc.15321] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022]
Abstract
Social rewards are fundamental to survival and overall health. Several studies suggest that adequate social stimuli during early life are critical for developing appropriate socioemotional and cognitive skills, whereas adverse social experiences negatively affect the proper development of brain and behavior, by increasing the susceptibility to develop neuropsychiatric conditions. Therefore, a better understanding of the neural mechanisms underlying social interactions, and their rewarding components in particular, is an important challenge of current neuroscience research. In this context, preclinical research has a crucial role: Animal models allow to investigate the neurobiological aspects of social reward in order to shed light on possible neurochemical alterations causing aberrant social reward processing in neuropsychiatric diseases, and they allow to test the validity and safety of innovative therapeutic strategies. Here, we discuss preclinical research that has investigated the rewarding properties of two forms of social interaction that occur in different phases of the lifespan of mammals, that is, mother-infant interaction and social interactions with peers, by focusing on the main neurotransmitter systems mediating their rewarding components. Together, the research performed so far helped to elucidate the mechanisms of social reward and its psychobiological components throughout development, thus increasing our understanding of the neurobiological substrates sustaining social functioning in health conditions and social dysfunction in major psychiatric disorders.
Collapse
Affiliation(s)
- Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy.,Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Emilia Carbone
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Claudia Cacchione
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy.,Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| |
Collapse
|
33
|
Xu P, Lai M, Fu D, Liu H, Wang Y, Shen H, Zhou W. Reinforcing and discriminative-stimulus effects of two pyrrolidine-containing synthetic cathinone derivatives in rats. Pharmacol Biochem Behav 2021; 203:173128. [PMID: 33515585 DOI: 10.1016/j.pbb.2021.173128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 11/30/2022]
Abstract
The molecular and behavioral aspects of α-pyrrolidinopentiophenone (α-PVP) have been characterized; however, how the structural modification of α-PVP affects its abuse potential is still unknown. In this study, we investigated the abuse potential of two pyrrolidinylated second-generation cathinones:4-chloro-α-pyrrolidinopentiophenone (4cl-α-PVP) and 4-chloro-α-pyrrolidinopropiophenone (4cl-α-PPP). Male Sprague-Dawley rats were trained to self-administer methamphetamine (METH, 0.05 mg·kg-1·infusion-1), α-PVP (0.05 mg·kg-1·infusion-1), 4cl-α-PVP (0.05 mg·kg-1·infusion-1), and 4cl-α-PPP (0.5 mg·kg-1·infusion-1) under a fixed ratio (FR) 1 reinforcement schedule for 10 sessions. The discriminative-stimulus effect of METH (0.8 mg/kg) from saline was tested under an FR10 schedule of food delivery. α-PVP, 4cl-α-PVP and 4cl-α-PPP produced reinforcement behaviors and presented an inverted U-shaped dose effect. The reinforcing potency was displayed with a rank order of α-PVP (0.029 mg·kg-1·infusion-1) > METH (0.040 mg·kg-1·infusion-1) > 4cl-α-PVP (0.094 mg·kg-1·infusion-1) > 4cl-α-PPP (0.51 mg·kg-1·infusion-1). All three drugs were fully substituted for the discriminative-stimulus effects of METH in rats. The substitution potency for discriminative-stimulus effects of α-PVP (ED50 = 0.4 mg/kg) was approximately equal to that of METH (ED50 = 0.3 mg/kg), while the discriminative potency of 4cl-α-PVP (ED50 = 1.0 mg/kg) and 4cl-α-PPP (ED50 = 5 mg/kg) was approximately 3 and 16-fold less than that of METH. The rank order of potency was α-PVP ≈ METH >4cl-α-PVP > 4cl-α-PPP. The present data demonstrated that 4cl-α-PVP and 4cl-α-PPP produced reinforcing effects and fully and dose-dependently substituted for the subjective effects of METH, suggesting that both 4cl-α-PVP and 4cl-α-PPP have abuse potential that may be similar to METH.
Collapse
Affiliation(s)
- Peng Xu
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100093, PR China
| | - Miaojun Lai
- Zhejiang Provincial Key Laboratory of Addiction, Ningbo Kangning Hospital, School of Medicine, Ningbo University, Ningbo 315201, Zhejiang Province, PR China
| | - Dan Fu
- Zhejiang Provincial Key Laboratory of Addiction, Ningbo Kangning Hospital, School of Medicine, Ningbo University, Ningbo 315201, Zhejiang Province, PR China
| | - Huifen Liu
- Zhejiang Provincial Key Laboratory of Addiction, Ningbo Kangning Hospital, School of Medicine, Ningbo University, Ningbo 315201, Zhejiang Province, PR China
| | - Youmei Wang
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100093, PR China
| | - Haowei Shen
- Faculty of Physiology & Pharmacology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang Province, PR China.
| | - Wenhua Zhou
- Zhejiang Provincial Key Laboratory of Addiction, Ningbo Kangning Hospital, School of Medicine, Ningbo University, Ningbo 315201, Zhejiang Province, PR China.
| |
Collapse
|
34
|
Townsend EA, Negus SS, Banks ML. Medications Development for Treatment of Opioid Use Disorder. Cold Spring Harb Perspect Med 2021; 11:a039263. [PMID: 31932466 PMCID: PMC7778216 DOI: 10.1101/cshperspect.a039263] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review describes methods for preclinical evaluation of candidate medications to treat opioid use disorder (OUD). The review is founded on the propositions that (1) drug self-administration procedures provide the most direct method for assessment of medication effectiveness, (2) procedures that assess choice between opioid and nondrug reinforcers are especially useful, and (3) states of opioid dependence and withdrawal profoundly influence both opioid reinforcement and effects of candidate medications. Effects of opioid medications and vaccines on opioid choice in nondependent and opioid-dependent subjects are reviewed. Various nonopioid medications have also been examined, but none yet have been identified that safely and reliably reduce opioid choice. Future research will focus on (1) strategies for increasing safety and/or effectiveness of opioid medications (e.g., G-protein-biased μ-opioid agonists), and (2) continued development of nonopioid medications (e.g., clonidine) that might serve as adjunctive agents to current opioid medications.
Collapse
Affiliation(s)
- E Andrew Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| |
Collapse
|
35
|
Clasen MM, Riley AL, Davidson TL. Hippocampal-Dependent Inhibitory Learning and Memory Processes in the Control of Eating and Drug Taking. Curr Pharm Des 2020; 26:2334-2352. [PMID: 32026771 DOI: 10.2174/1381612826666200206091447] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
As manifestations of excessive and uncontrolled intake, obesity and drug addiction have generated much research aimed at identifying common neuroadaptations that could underlie both disorders. Much work has focused on changes in brain reward and motivational circuitry that can overexcite eating and drug-taking behaviors. We suggest that the regulation of both behaviors depends on balancing excitation produced by stimuli associated with food and drug rewards with the behavioral inhibition produced by physiological "satiety" and other stimuli that signal when those rewards are unavailable. Our main hypothesis is that dysregulated eating and drug use are consequences of diet- and drug-induced degradations in this inhibitory power. We first outline a learning and memory mechanism that could underlie the inhibition of both food and drug-intake, and we describe data that identifies the hippocampus as a brain substrate for this mechanism. We then present evidence that obesitypromoting western diets (WD) impair the operation of this process and generate pathophysiologies that disrupt hippocampal functioning. Next, we present parallel evidence that drugs of abuse also impair this same learning and memory process and generate similar hippocampal pathophysiologies. We also describe recent findings that prior WD intake elevates drug self-administration, and the implications of using drugs (i.e., glucagon-like peptide- 1 agonists) that enhance hippocampal functioning to treat both obesity and addiction are also considered. We conclude with a description of how both WD and drugs of abuse could initiate a "vicious-cycle" of hippocampal pathophysiology and impaired hippocampal-dependent behavioral inhibition.
Collapse
Affiliation(s)
- Matthew M Clasen
- Department of Psychology, Program in Neuroscience, Williams College, Williamstown, MA 01267, United States
| | - Anthony L Riley
- Department of Neuroscience, Center for Behavioral Neuroscience, American University, Washington, DC 20016, United States
| | - Terry L Davidson
- Department of Neuroscience, Center for Behavioral Neuroscience, American University, Washington, DC 20016, United States
| |
Collapse
|
36
|
Transcriptional Profiling of Whisker Follicles and of the Striatum in Methamphetamine Self-Administered Rats. Int J Mol Sci 2020; 21:ijms21228856. [PMID: 33238484 PMCID: PMC7700365 DOI: 10.3390/ijms21228856] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Methamphetamine (MA) use disorder is a chronic neuropsychiatric disease characterized by recurrent binge episodes, intervals of abstinence, and relapses to MA use. Therefore, identification of the key genes and pathways involved is important for improving the diagnosis and treatment of this disorder. In this study, high-throughput RNA sequencing was performed to find the key genes and examine the comparability of gene expression between whisker follicles and the striatum of rats following MA self-administration. A total of 253 and 87 differentially expressed genes (DEGs) were identified in whisker follicles and the striatum, respectively. Multivariate and network analyses were performed on these DEGs to find hub genes and key pathways within the constructed network. A total of 129 and 49 genes were finally selected from the DEG sets of whisker follicles and of the striatum. Statistically significant DEGs were found to belong to the classes of genes involved in nicotine addiction, cocaine addiction, and amphetamine addiction in the striatum as well as in Parkinson’s, Huntington’s, and Alzheimer’s diseases in whisker follicles. Of note, several genes and pathways including retrograde endocannabinoid signaling and the synaptic vesicle cycle pathway were common between the two tissues. Therefore, this study provides the first data on gene expression levels in whisker follicles and in the striatum in relation to MA reward and thereby may accelerate the research on the whisker follicle as an alternative source of biomarkers for the diagnosis of MA use disorder.
Collapse
|
37
|
McClenahan SJ, Gunnell MG, Owens SM, Fantegrossi WE. Active vaccination reduces reinforcing effects of MDPV in male Sprague-Dawley rats trained to self-administer cocaine. Psychopharmacology (Berl) 2020; 237:2613-2620. [PMID: 32500210 PMCID: PMC7502518 DOI: 10.1007/s00213-020-05558-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
RATIONALE 3,4-Methylenedioxypyrovalerone (MDPV) is a synthetic cathinone abused for its cocaine-like psychostimulant effects in "bath salts" products. While there are currently no pharmacotherapies for MDPV abuse, rodent studies suggest immunotherapy may offer a feasible treatment option. OBJECTIVES These studies tested the capacity of active vaccination to reduce the reinforcing effects of MDPV in Sprague-Dawley rats. METHODS Rats acquired cocaine self-administration (0.32 mg/kg/inf) on an FR1 schedule. Dose-effect functions for cocaine (0.032-1.0 mg/kg/inf) and MDPV (0.001-0.32 mg/kg/inf) were determined under an FR5 schedule. Rats in the vaccine group were immunized during cocaine self-administration. All rats transitioned to a progressive-ratio (PR) schedule to establish breakpoints for cocaine (0.1-1.0 mg/kg/inf) and MDPV (0.01-0.32 mg/kg/inf). Responding was extinguished, and cue-induced and MDPV-primed reinstatement (0.56 mg/kg, IP) were evaluated. RESULTS No endpoints of cocaine self-administration differed between groups, but the ED50 for MDPV self-administration was significantly lower in control relative to vaccinated rats. Under the PR schedule, MDPV was ~ 2.5-fold more potent in maintaining responding in control than vaccinated rats, but Emax was not different between groups. Vaccination did not reduce MDPV-primed reinstatement, perhaps due to a decrease in antibody titer. CONCLUSIONS Vaccination did not alter acquisition of cocaine self-administration, demonstrating pharmacological selectivity and suggesting that the vaccine did not affect learning or motivation, while effectively reducing the potency of MDPV as a reinforcer. The protective effects of the vaccine were surmounted by large unit doses of MDPV, suggesting maximal efficacy of drug-conjugate vaccines in substance abuse disorders will likely require concurrent behavior modification therapy.
Collapse
Affiliation(s)
- Samantha J McClenahan
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham, Slot 638, Little Rock, AR, 72205, USA
| | - Melinda G Gunnell
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham, Slot 638, Little Rock, AR, 72205, USA
| | - S Michael Owens
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham, Slot 638, Little Rock, AR, 72205, USA
| | - William E Fantegrossi
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham, Slot 638, Little Rock, AR, 72205, USA.
| |
Collapse
|
38
|
Windisch KA, Kreek MJ. Review of addiction risk potential associated with adolescent opioid use. Pharmacol Biochem Behav 2020; 198:173022. [PMID: 32871141 DOI: 10.1016/j.pbb.2020.173022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 01/31/2023]
Abstract
Adolescence is a critical period of development with robust behavioral, morphological, hormonal, and neurochemical changes including changes in brain regions implicated in the reinforcing effects of drugs such as opioids. Here we examine the preclinical and, where appropriate complementary clinical literature, for the behavioral and neurological changes induced by adolescent opioid exposure/use and their long-term consequences during adulthood. Adolescent opioid exposure results in a widened biphasic shift in reinforcement with increased impact of positive rewarding aspects during initial use and profound negative reinforcement during adulthood. Females may have enhanced vulnerability due to fast onset of antinociceptive tolerance and reduced severity of somatic withdrawal symptoms during adolescence. Overall, adolescent opioid exposure, be it legally prescribed protracted intake or illicit consumption, results in significant and prolonged consequences of increased opioid reward concomitant with reduced analgesic efficacy and exacerbated somatic withdrawal severity during opioid use/exposure in adulthood. These findings are highly relevant to physicians, parents, law makers, and the general public as adolescent opioid exposure/misuse results in heightened risk for substance use disorders.
Collapse
Affiliation(s)
- Kyle A Windisch
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
39
|
The kappa-opioid receptor agonist, nalfurafine, blocks acquisition of oxycodone self-administration and oxycodone's conditioned rewarding effects in male rats. Behav Pharmacol 2020; 31:792-797. [PMID: 32804774 DOI: 10.1097/fbp.0000000000000581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mu-opioid receptor (MOR) agonists are highly efficacious for the treatment of pain but have significant abuse liability. Recently, we reported that nalfurafine, when combined with oxycodone at a certain ratio, reduced the reinforcing effects of oxycodone in rats while producing additive antinociceptive effects. Questions remain, however, including if the combination will function as a reinforcer in drug-naïve rats, and if the combination produces aversive effects that could explain nalfurafine's ability to reduce oxycodone self-administration? In the present study, we investigated nalfurafine's ability to reduce acquisition of oxycodone self-administration when the two were self-administered as a mixture in drug-naïve rats and nalfurafine's ability to attenuate a conditioned place preference (CPP) induced by oxycodone. In the self-administration study, male Sprague-Dawley rats self-administered intravenous injections of oxycodone (0.056 mg/kg/injection), an oxycodone/nalfurafine combination (0.056/0.0032 mg/kg/injection), or saline under fixed-ratio schedules of reinforcement for 20 days to compare rates of acquisition of drug taking. In the CPP assay, male Sprague-Dawley rats received subcutaneous injections of either saline, oxycodone (3.2 mg/kg), nalfurafine (0.18 mg/kg), or an oxycodone/nalfurafine combination at the same ratio used in the self-administration study (3.2 mg/kg/0.18 mg/kg). All subjects self-administering oxycodone alone met acquisition criteria. However, only 13% of subjects self-administering oxycodone/nalfurafine met criteria, and no subjects acquired self-administration of saline. Oxycodone, but not nalfurafine alone or the oxycodone/nalfurafine combination, produced rewarding effects in rats in the CPP test. These findings suggest that the combination of oxycodone and nalfurafine will be less habit forming in opioid-naïve patients than oxycodone alone.
Collapse
|
40
|
Cabañero D, Ramírez-López A, Drews E, Schmöle A, Otte DM, Wawrzczak-Bargiela A, Huerga Encabo H, Kummer S, Ferrer-Montiel A, Przewlocki R, Zimmer A, Maldonado R. Protective role of neuronal and lymphoid cannabinoid CB 2 receptors in neuropathic pain. eLife 2020; 9:55582. [PMID: 32687056 PMCID: PMC7384863 DOI: 10.7554/elife.55582] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/19/2020] [Indexed: 12/14/2022] Open
Abstract
Cannabinoid CB2 receptor (CB2) agonists are potential analgesics void of psychotropic effects. Peripheral immune cells, neurons and glia express CB2; however, the involvement of CB2 from these cells in neuropathic pain remains unresolved. We explored spontaneous neuropathic pain through on-demand self-administration of the selective CB2 agonist JWH133 in wild-type and knockout mice lacking CB2 in neurons, monocytes or constitutively. Operant self-administration reflected drug-taking to alleviate spontaneous pain, nociceptive and affective manifestations. While constitutive deletion of CB2 disrupted JWH133-taking behavior, this behavior was not modified in monocyte-specific CB2 knockouts and was increased in mice defective in neuronal CB2 knockouts suggestive of increased spontaneous pain. Interestingly, CB2-positive lymphocytes infiltrated the injured nerve and possible CB2transfer from immune cells to neurons was found. Lymphocyte CB2depletion also exacerbated JWH133 self-administration and inhibited antinociception. This work identifies a simultaneous activity of neuronal and lymphoid CB2that protects against spontaneous and evoked neuropathic pain.
Collapse
Affiliation(s)
- David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Angela Ramírez-López
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eva Drews
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Anne Schmöle
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - David M Otte
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Agnieszka Wawrzczak-Bargiela
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Hector Huerga Encabo
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Sami Kummer
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Antonio Ferrer-Montiel
- Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
41
|
Huskinson SL. Unpredictability as a modulator of drug self-administration: Relevance for substance-use disorders. Behav Processes 2020; 178:104156. [PMID: 32526314 DOI: 10.1016/j.beproc.2020.104156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/14/2020] [Accepted: 05/29/2020] [Indexed: 01/05/2023]
Abstract
Drug self-administration has been regarded as a gold-standard preclinical model of addiction and substance-use disorder (SUD). However, investigators are becoming increasingly aware, that certain aspects of addiction or SUDs experienced by humans are not accurately captured in our preclinical self-administration models. The current review will focus on two such aspects of current preclinical drug self-administration models: 1) Predictable vs. unpredictable drug access in terms of the time and effort put into obtaining drugs (i.e., response requirement) and drug quality (i.e., amount) and 2) rich vs. lean access to drugs. Some behavioral and neurobiological mechanisms that could contribute to excessive allocation of behavior toward drug-seeking and drug-taking at the expense of engaging in nondrug-related activities are discussed, and some directions for future research are identified. Based on the experiments reviewed, lean and unpredictable drug access could worsen drug-seeking and drug-taking behavior in individuals with SUDs. Once more fully explored, this area of research will help determine whether and how unpredictable and lean cost requirements affect drug self-administration in preclinical laboratory studies with nonhuman subjects and will help determine whether incorporating these conditions in current self-administration models will increase their predictive validity.
Collapse
Affiliation(s)
- Sally L Huskinson
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500N. State Street, Jackson, MS, 39216, United States.
| |
Collapse
|
42
|
Green TA, Bardo MT. Opposite regulation of conditioned place preference and intravenous drug self-administration in rodent models: Motivational and non-motivational examples. Neurosci Biobehav Rev 2020; 116:89-98. [PMID: 32534899 DOI: 10.1016/j.neubiorev.2020.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Abstract
Although developed from a common antecedent, conditioned place preference (CPP) and intravenous drug self-administration (SA) represent different behavioral paradigms, each with strong face validity. The field has treated results from these studies largely interchangeably; however, there is considerable evidence of opposite modulation of CPP vs. SA. This review outlines four manipulations that differentially affect CPP and SA based on alterations of motivation. These examples are contrasted with one example of differential CPP and SA results that can be explained by simple parallel shifts in dose-response functions. The final two examples have yet to be classified as motivation-based or parallel shifts. Important aspects, including motivation, volitional control of drug administration, reward, and the role of cues are discussed. One major conclusion of this paper is that explanations for apparent discrepancies between CPP and SA require full dose effect functions and assessment of PR breakpoints. Overall, this manuscript offers a more nuanced insight into how CPP and SA can be used to study different aspects of substance use disorders.
Collapse
Affiliation(s)
- Thomas A Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, United States.
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, United States.
| |
Collapse
|
43
|
Riley AL, Nelson KH, To P, López-Arnau R, Xu P, Wang D, Wang Y, Shen HW, Kuhn DM, Angoa-Perez M, Anneken JH, Muskiewicz D, Hall FS. Abuse potential and toxicity of the synthetic cathinones (i.e., “Bath salts”). Neurosci Biobehav Rev 2020; 110:150-173. [DOI: 10.1016/j.neubiorev.2018.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/14/2018] [Accepted: 07/24/2018] [Indexed: 01/22/2023]
|
44
|
Nonhuman animal models of substance use disorders: Translational value and utility to basic science. Drug Alcohol Depend 2020; 206:107733. [PMID: 31790978 PMCID: PMC6980671 DOI: 10.1016/j.drugalcdep.2019.107733] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The National Institute on Drug Abuse (NIDA) recently released a Request for Information (RFI) soliciting comments on nonhuman animal models of substance use disorders (SUD). METHODS A literature review was performed to address the four topics outlined in the RFI and one topic inspired by the RFI: (1) animal models that best recapitulate SUD, (2) animal models that best balance the trade-offs between resources and ecological validity, (3) animal models whose translational value are frequently misrepresented or overrepresented by the scientific community, (4) aspects of SUD that are not currently being modeled in animals, and (5) animal models that are optimal for examining the basic mechanisms by which drugs produce their abuse-related effects. RESULTS Models that employ response-contingent drug administration, use complex schedules of reinforcement, measure behaviors that mimic the distinguishing features of SUD, and use animals that are phylogenetically similar to humans have the greatest translational value. Models that produce stable and reproducible baselines of behavior, lessen the number of uncontrolled variables, and minimize the influence of extraneous factors are best at examining basic mechanisms contributing to drug reward and reinforcement. CONCLUSIONS Nonhuman animal models of SUD have undergone significant refinements to increase their utility for basic science and translational value for SUD. The existing literature describes numerous examples of how these models may best be utilized to answer mechanistic questions of drug reward and identify potential therapeutic interventions for SUD. Progress in the field could be accelerated by further collaborations between researchers using animals versus humans.
Collapse
|
45
|
Sartor GC. Epigenetic pharmacotherapy for substance use disorder. Biochem Pharmacol 2019; 168:269-274. [PMID: 31306644 PMCID: PMC6733674 DOI: 10.1016/j.bcp.2019.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/10/2019] [Indexed: 12/12/2022]
Abstract
Identifying novel therapeutics for the treatment of substance use disorder (SUD) is an area of intensive investigation. Prior strategies that have attempted to modify one or a few neurotransmitter receptors have had limited success, and currently there are no FDA-approved medications for the treatment of cocaine, methamphetamine, and marijuana use disorders. Because drugs of abuse are known to alter the expression of numerous genes in reward-related brain regions, epigenetic-based therapies have emerged as intriguing targets for therapeutic innovation. Here, I evaluate potential therapeutic approaches and challenges in targeting epigenetic factors for the treatment of SUD and highlight examples of promising strategies and future directions.
Collapse
Affiliation(s)
- Gregory C Sartor
- University of Connecticut, Department of Pharmaceutical Sciences, 69 N. Eagleville Road, Storrs, CT 06269, United States.
| |
Collapse
|
46
|
Hemby SE, McIntosh S, Leon F, Cutler SJ, McCurdy CR. Abuse liability and therapeutic potential of the Mitragyna speciosa (kratom) alkaloids mitragynine and 7-hydroxymitragynine. Addict Biol 2019; 24:874-885. [PMID: 29949228 DOI: 10.1111/adb.12639] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/20/2018] [Accepted: 05/10/2018] [Indexed: 12/17/2022]
Abstract
Kratom, derived from the plant Mitragyna speciosa, is receiving increased attention as an alternative to traditional opiates and as a replacement therapy for opiate dependence. Mitragynine (MG) and 7-hydroxymitragynine (7-HMG) are major psychoactive constituents of kratom. While MG and 7-HMG share behavioral and analgesic properties with morphine, their reinforcing effects have not been examined to date. 7-HMG, but not MG, substituted for morphine self-administration in a dose-dependent manner in the rat self-administration paradigm. Following the substitution procedure, re-assessment of morphine self-administration revealed a significant increase following 7-HMG and a significant decrease following MG substitution. In a separate cohort, 7-HMG, but not MG, engendered and maintained intravenous self-administration in a dose-dependent manner. The effects of pretreatment with nalxonaxine (NLXZ), a μ1 opiate receptor antagonist, and naltrindole (NTI), a δ opiate receptor antagonist, on 7-HMG and morphine self-administration were also examined. Both NLXZ and NTI reduced 7-HMG self-administration, whereas only NLXZ decreased morphine intake. The present results are the first to demonstrate that 7-HMG is readily self-administered, and the reinforcing effects of 7-HMG are mediated in part by μ and δ opiate receptors. In addition, prior exposure to 7-HMG increased subsequent morphine intake whereas prior exposure to MG decreased morphine intake. The present findings indicate that MG does not have abuse potential and reduces morphine intake, desired characteristics of candidate pharmacotherapies for opiate addiction and withdrawal, whereas 7-HMG should be considered a kratom constituent with high abuse potential that may also increase the intake of other opiates.
Collapse
Affiliation(s)
- Scott E. Hemby
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy; High Point University; High Point NC USA
| | - Scot McIntosh
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy; High Point University; High Point NC USA
| | - Francisco Leon
- Department of Medicinal Chemistry, College of Pharmacy; University of Florida; Gainesville FL USA
| | | | - Christopher R. McCurdy
- Department of Medicinal Chemistry, College of Pharmacy; University of Florida; Gainesville FL USA
| |
Collapse
|
47
|
Abstract
All preclinical procedures for analgesic drug discovery involve two components: 1) a "pain stimulus" (the principal independent variable), which is delivered to an experimental subject with the intention of producing a pain state; and 2) a "pain behavior" (the principal dependent variable), which is measured as evidence of that pain state. Candidate analgesics are then evaluated for their effectiveness to reduce the pain behavior, and results are used to prioritize drugs for advancement to clinical testing. This review describes a taxonomy of preclinical procedures organized into an "antinociception matrix" by reference to their types of pain stimulus (noxious, inflammatory, neuropathic, disease related) and pain behavior (unconditioned, classically conditioned, operant conditioned). Particular emphasis is devoted to pain behaviors and the behavioral principals that govern their expression, pharmacological modulation, and preclinical-to-clinical translation. Strengths and weaknesses are compared and contrasted for procedures using each type of behavioral outcome measure, and the following four recommendations are offered to promote strategic use of these procedures for preclinical-to-clinical analgesic drug testing. First, attend to the degree of homology between preclinical and clinical outcome measures, and use preclinical procedures with behavioral outcome measures homologous to clinically relevant outcomes in humans. Second, use combinations of preclinical procedures with complementary strengths and weaknesses to optimize both sensitivity and selectivity of preclinical testing. Third, take advantage of failed clinical translation to identify drugs that can be back-translated preclinically as active negative controls. Finally, increase precision of procedure labels by indicating both the pain stimulus and the pain behavior in naming preclinical procedures.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
48
|
Smith SL, Dean RL, Todtenkopf MS, Heal DJ. Investigation of the reinforcing potential of samidorphan and naltrexone by fixed and progressive ratio intravenous self-administration testing in heroin-maintained rats. J Psychopharmacol 2019; 33:383-391. [PMID: 30676189 DOI: 10.1177/0269881118822111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Samidorphan is a novel μ-opioid antagonist with low intrinsic activity at κ- and δ-opioid receptors. AIMS Because samidorphan is central nervous system-active, we investigated whether samidorphan (13.6, 40.8, 68 μg/kg/injection) served as a positive reinforcer in rats trained to self-administer heroin on a fixed ratio-5 schedule. Samidorphan's relative reinforcing effect was evaluated by progressive ratio/break-point determination. Naltrexone (13.6, 40.8, 68 μg/kg/injection) and heroin (7.5, 15, 25 μg/kg/injection) were comparators. RESULTS All heroin doses maintained self-administration on fixed ratio-5 and progressive ratio/break-points at levels significantly greater than saline. Samidorphan and naltrexone had similar profiles on fixed ratio-5 with one samidorphan dose serving as a positive reinforcer and one naltrexone dose showing a strong trend ( p=0.053) for positive reinforcement. The numbers of injections of every samidorphan and naltrexone dose were significantly lower than all heroin doses. The numbers of self-administered samidorphan and naltrexone injections/session on fixed ratio-5 were not significantly different from one another. The mean inter-injection intervals for heroin were significantly shorter than for saline, whereas those of samidorphan and naltrexone were not. Progressive ratio break-points for samidorphan and naltrexone were not different from saline except for the highest dose of samidorphan. In addition, the progressive ratio break-points for samidorphan were not significantly different from those of naltrexone and were significantly lower than heroin. The samidorphan unit-doses evaluated in self-administration yielded plasma concentrations ranging between 25-109% and 10-45% of the maximum concentration values in humans. CONCLUSIONS Overall, the profiles of samidorphan and naltrexone, which has no abuse liability, were similar in this model.
Collapse
|
49
|
Jeon SY, Kim YH, Kim SJ, Suh SK, Cha HJ. Abuse potential of 2-(4-iodo-2, 5-dimethoxyphenyl)N-(2-methoxybenzyl)ethanamine (25INBOMe); in vivo and ex vivo approaches. Neurochem Int 2019; 125:74-81. [PMID: 30769030 DOI: 10.1016/j.neuint.2019.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/10/2019] [Accepted: 02/12/2019] [Indexed: 01/08/2023]
Abstract
25INBOMe ("25-I", "N-Bomb"), one of new psychoactive substances (NPSs), is being abused for recreational purpose. However, the liability for abuse or dependence has not been systematically studied yet. The objective of the present study was to evaluate rewarding and reinforcing effects of 25INBOMe using conditioned place preference (CPP) and self-administration (SA) paradigms. In addition, ultrasonic vocalizations (USVs) were measured to investigate relationships between USVs and emotional state regarding dependence on psychoactive substances. To understand molecular mechanism involved in its action, dopamine (DA) level changes were analyzed using synaptosomes extracted from the striatal region of the brain. Expression level changes of SGK1 (serum/glucocorticoid regulated kinase 1) and PER2 (period circadian protein homolog 2), two putative biomarkers for drug dependence, were also analyzed. Results showed that 25INBOMe increased both CPP (0.3 mg/kg) and SA (0.03 mg/kg/infusion) and produced higher frequencies in USVs analysis. It also increased DA levels in the striatal region and changed expression levels of SGK1 and PER2. Results of the present study suggest that 25INBOMe might produce rewarding and reinforcing effects, indicating its dependence liability. In addition, frequencies of USV might be associated with emotional state of mice induced by psychoactive substances regarding substance dependence. This is the first systemic preclinical report on the dependence liability of 25INBOMe and the first attempt to introduce a possible relationship between USVs and emotional state of mice regarding substance dependency. Further studies are needed to clarify the mechanism involved in 25INBOMe dependency and determine the usefulness of USV measurement as a method for evaluating dependence liability.
Collapse
Affiliation(s)
- Seo Young Jeon
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Young-Hoon Kim
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Sung Jin Kim
- Cosmetics Policy Division, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Soo Kyung Suh
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Hye Jin Cha
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| |
Collapse
|
50
|
Atigari DV, Uprety R, Pasternak GW, Majumdar S, Kivell BM. MP1104, a mixed kappa-delta opioid receptor agonist has anti-cocaine properties with reduced side-effects in rats. Neuropharmacology 2019; 150:217-228. [PMID: 30768946 DOI: 10.1016/j.neuropharm.2019.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/23/2019] [Accepted: 02/09/2019] [Indexed: 01/02/2023]
Abstract
Kappa opioid receptor (KOPr) agonists have preclinical anti-cocaine and antinociceptive effects. However, adverse effects including dysphoria, aversion, sedation, anxiety and depression limit their clinical development. MP1104, an analogue of 3-iodobenzoyl naltrexamine, is a potent dual agonist at KOPr and delta opioid receptor (DOPr), with full agonist efficacy at both these receptors. In this study, we evaluate the ability of MP1104 to modulate cocaine-induced behaviors and side-effects preclinically. In male Sprague-Dawley rats trained to self-administer cocaine, MP1104 (0.3 and 1 mg/kg) reduced cocaine-primed reinstatement of drug-seeking behavior and caused significant downward shift of the dose-response curve in cocaine self-administration tests (0.3 and 0.6 mg/kg). The anti-cocaine effects exerted by MP1104 are in part due to increased dopamine (DA) uptake by the dopamine transporter (DAT) in the dorsal striatum (dStr) and nucleus accumbens (NAc). MP1104 (0.3 and 0.6 mg/kg) showed no significant anxiogenic effects in the elevated plus maze, pro-depressive effects in the forced swim test, or conditioned place aversion. Furthermore, pre-treatment with a DOPr antagonist, led to MP1104 producing aversive effects. This data suggests that the DOPr agonist actions of MP1104 attenuate the KOPr-mediated aversive effects of MP1104. The overall results from this study show that MP1104, modulates DA uptake in the dStr and NAc, and exerts potent anti-cocaine properties in self-administration tests with reduced side-effects compared to pure KOPr agonists. This data supports the therapeutic development of dual KOPr/DOPr agonists to reduce the side-effects of selective KOPr agonists. This article is part of the Special Issue entitled 'Opioid Neuropharmacology: Advances in treating pain and opioid addiction'.
Collapse
Affiliation(s)
- Diana V Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Rajendra Uprety
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - Gavril W Pasternak
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - Susruta Majumdar
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA; Center for Clinical Pharmacology, St Louis College of Pharmacy and Washington University School of Medicine, St Louis, MO, USA
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|